Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 94
Filter
1.
Exp Eye Res ; 219: 109081, 2022 06.
Article in English | MEDLINE | ID: mdl-35461874

ABSTRACT

The human cornea is responsible for approximately 70% of the eye's optical power and, together with the lens, constitutes the only transparent tissue in the human body. Low-density lipoprotein receptor-related protein 1 (LRP1), a large, multitalented endocytic receptor, is expressed throughout the human cornea, yet its role in the cornea remains unknown. More than 30 years ago, LRP1 was purified by exploiting its affinity for the activated form of the protease inhibitor alpha-2-macroblulin (A2M), and the original purification protocol is generally referred to in studies involving full-length LRP1. Here, we provide a novel and simplified LRP1 purification protocol based on LRP1's affinity for receptor-related protein (RAP) that produces significantly higher yields of authentic LRP1. Purified LRP1 was used to map its unknown interactome in the human cornea. Corneal proteins extracted under physiologically relevant conditions were subjected to LRP1 affinity pull-down, and LRP1 ligand candidates were identified by LC-MS/MS. A total of 28 LRP1 ligand candidates were found, including 22 novel ligands. The LRP1 corneal interactome suggests a novel role for LRP1 as a regulator of the corneal immune response, structure, and ultimately corneal transparency.


Subject(s)
Cornea , Low Density Lipoprotein Receptor-Related Protein-1 , Protein Interaction Mapping , Chromatography, Liquid , Cornea/chemistry , Cornea/metabolism , Humans , Ligands , Lipoproteins, LDL , Low Density Lipoprotein Receptor-Related Protein-1/chemistry , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Protein Interaction Mapping/methods , Tandem Mass Spectrometry
2.
J Biol Chem ; 297(1): 100842, 2021 07.
Article in English | MEDLINE | ID: mdl-34058195

ABSTRACT

The low-density lipoprotein receptor (LDLR) family of receptors are cell-surface receptors that internalize numerous ligands and play crucial role in various processes, such as lipoprotein metabolism, hemostasis, fetal development, etc. Previously, receptor-associated protein (RAP) was described as a molecular chaperone for LDLR-related protein 1 (LRP1), a prominent member of the LDLR family. We aimed to verify this role of RAP for LRP1 and two other LDLR family receptors, LDLR and vLDLR, and to investigate the mechanisms of respective interactions using a cell culture model system, purified system, and in silico modelling. Upon coexpression of RAP with clusters of the ligand-binding complement repeats (CRs) of the receptors in secreted form in insect cells culture, the isolated proteins had increased yield, enhanced folding, and improved binding properties compared with proteins expressed without RAP, as determined by circular dichroism and surface plasmon resonance. Within LRP1 CR-clusters II and IV, we identified multiple sites comprised of adjacent CR doublets, which provide alternative bivalent binding combinations with specific pairs of lysines on RAP. Mutational analysis of these lysines within each of isolated RAP D1/D2 and D3 domains having high affinity to LRP1 and of conserved tryptophans on selected CR-doublets of LRP1, as well as in silico docking of a model LRP1 CR-triplet with RAP, indicated a universal role for these residues in interaction of RAP and LRP1. Consequently, we propose a new model of RAP interaction with LDLR family receptors based on switching of the bivalent contacts between molecules over time in a dynamic mode.


Subject(s)
LDL-Receptor Related Protein-Associated Protein/metabolism , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Protein Folding , Receptors, LDL/metabolism , DNA Mutational Analysis , Humans , Ligands , Low Density Lipoprotein Receptor-Related Protein-1/chemistry , Molecular Docking Simulation , Protein Binding , Repetitive Sequences, Amino Acid
3.
Theranostics ; 10(7): 3263-3280, 2020.
Article in English | MEDLINE | ID: mdl-32194867

ABSTRACT

Background: The LRP1 (CR9) domain and, in particular, the sequence Gly1127-Cys1140 (P3) plays a critical role in the binding and internalization of aggregated LDL (agLDL). We aimed to evaluate whether immunization with P3 reduces high-fat diet (HFD)-induced atherosclerosis. Methods: Female New Zealand White (NZW) rabbits were immunized with a primary injection and four reminder doses (R1-R4) of IrP (irrelevant peptide) or P3 conjugated to the carrier. IrP and P3-immunized rabbits were randomly divided into a normal diet group and a HFD-fed group. Anti-P3 antibody levels were determined by ELISA. Lipoprotein profile, circulating and tissue lipids, and vascular pro-inflammatory mediators were determined using standardized methods while atherosclerosis was determined by confocal microscopy studies and non-invasive imaging (PET/CT and Doppler ultrasonography). Studies treating human macrophages (hMΦ) and coronary vascular smooth muscle cells (hcVSMC) with rabbit serums were performed to ascertain the potential impact of anti-P3 Abs on the functionality of these crucial cells. Results: P3 immunization specifically induced the production of anti-P3 antibodies (Abs) and did not alter the lipoprotein profile. HFD strongly induced cholesteryl ester (CE) accumulation in the aorta of both the control and IrP groups, and their serum dose-dependently raised the intracellular CE of hMΦ and hcVSMC, promoting TNFR1 and phospho-NF-kB (p65) overexpression. These HFD pro-inflammatory effects were dramatically decreased in the aorta of P3-immunized rabbits and in hMΦ and hcVSMC exposed to the P3 rabbit serums. Microscopy studies revealed that P3 immunization reduced the percentage of lipids, macrophages, and SMCs in the arterial intima, as well as the atherosclerotic extent and lesion area in the aorta. PET/CT and Doppler ultrasonography studies showed that the average standardized uptake value (SUVmean) of the aorta and the arterial resistance index (ARI) of the carotids were more upregulated by HFD in the control and IrP groups than the P3 group. Conclusions: P3 immunization counteracts HFD-induced fatty streak formation in rabbits. The specific blockade of the LRP1 (CR9) domain with Anti-P3 Abs dramatically reduces HFD-induced intracellular CE loading and harmful coupling to pro-inflammatory signaling in the vasculature.


Subject(s)
Atherosclerosis/prevention & control , Immunization , Low Density Lipoprotein Receptor-Related Protein-1/immunology , Peptide Fragments/immunology , Amino Acid Sequence , Animals , Antibody Specificity , Aorta/cytology , Aorta/diagnostic imaging , Atherosclerosis/blood , Atherosclerosis/diagnostic imaging , Atherosclerosis/immunology , Cells, Cultured , Cholesterol Esters/metabolism , Coronary Vessels/cytology , Diet, High-Fat , Female , Humans , Lipids/blood , Lipoproteins/blood , Low Density Lipoprotein Receptor-Related Protein-1/chemistry , Macrophages/drug effects , Myocytes, Smooth Muscle/drug effects , Positron Emission Tomography Computed Tomography , Protein Domains , Rabbits , Random Allocation , Ultrasonography, Doppler , Vascular Resistance
4.
Nanotheranostics ; 3(4): 311-330, 2019.
Article in English | MEDLINE | ID: mdl-31687320

ABSTRACT

Glioblastoma is one of the most common primary tumor types of central nervous system (CNS) with high malignance and lethality. Although many treatment options are currently available, the therapy of brain cancers remains challenging because of blood-brain-barrier (BBB) which prevents most of the chemotherapeutics into the CNS. In this work, a poly(amidoamine) dendrimer-based carrier was fabricated and modified with angiopep-2 (Ang2) peptide that has been demonstrated to bind to low density lipoprotein receptor-relative protein-1 (LRP1) on the endothelial cells of BBB and could therefore induce BBB penetration of the carrier. To improve tumor-targeting effect towards the glioma sites, the dendrimer was simultaneously functionalized with an epidermal growth factor receptor (EGFR)-targeting peptide (EP-1) which was screened from a "one-bead one-compound" (OBOC) combinatorial library. EP-1 peptide was demonstrated to have high affinity and specificity to EGFR at both the molecular and cellular levels. The dual-targeting dendrimer exhibited outstanding BBB penetrability and glioma targeting efficiency both in vitro and in vivo, which strikingly enhanced the anti-gliomas effect of the drugs and prolonged the survival of gliomas-bearing mice. These results show the potential of the dual-targeting dendrimer-based carrier in the therapy of gliomas through enhancing BBB penetrability and tumor targeting.


Subject(s)
Blood-Brain Barrier , Dendrimers/chemistry , Peptides/chemistry , Polyamines/chemistry , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain Neoplasms/drug therapy , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Doxorubicin/chemistry , Doxorubicin/metabolism , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Female , Glioma/drug therapy , Glioma/mortality , Glioma/pathology , Humans , Kaplan-Meier Estimate , Low Density Lipoprotein Receptor-Related Protein-1/chemistry , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Mice , Mice, Nude , Peptides/metabolism , Protein Binding
5.
Cell Prolif ; 52(5): e12664, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31343104

ABSTRACT

OBJECTIVES: Low back pain becomes a common orthopaedic disease today. It is mainly induced by the degeneration of the intervertebral disc. In this study, we tried to reveal the pathogenesis of the degeneration and the relative therapeutic strategy, which are still elusive. MATERIALS AND METHODS: We collected 15 degenerative intervertebral tissues and five healthy donors. Nucleus pulposus and annulus fibrosus cells were subcultured. miR-640 expression was determined by qPCR. Computer analysis and luciferase reporter assay were used to confirm miR-640 target genes. Immunohistochemical and immunocytochemical staining was used to trace the proinflammatory cytokines and key transductor of signalling pathways. We also used ß-galactosidase staining, flow cytometry, and cell viability assay to monitor the degenerative index. RESULTS: miR-640 overexpressed in patients derived degenerative nucleus pulposus tissues and cells. The inflammatory environment promoted miR-640 expression via NF-κB signalling pathway. In addition, miR-640 targeted to LRP1 and enhances NF-κB signal activity, which built a positive feedback loop. miR-640 inhibited the expression of ß-catenin and EP300, therefore, restrained WNT signal and induced the degeneration in nucleus pulposus cells. miR-640 inhibitor treatment exhibited the effects of anti-inflammation, reverse WNT signalling pathway exhaustion, and remission of degenerative characteristics in vitro. CONCLUSIONS: miR-640 plays an important role in the degeneration of intervertebral disc and the relative inflammatory microenvironment. It is a promising potential therapeutic target for the low back pain biotherapy.


Subject(s)
Intervertebral Disc Degeneration/pathology , MicroRNAs/metabolism , NF-kappa B/metabolism , Signal Transduction , Wnt Proteins/metabolism , Adolescent , Adult , Annulus Fibrosus/cytology , Annulus Fibrosus/metabolism , Antagomirs/metabolism , Case-Control Studies , Cells, Cultured , E1A-Associated p300 Protein/metabolism , Humans , Interleukin-1beta/metabolism , Intervertebral Disc Degeneration/metabolism , Low Density Lipoprotein Receptor-Related Protein-1/chemistry , Low Density Lipoprotein Receptor-Related Protein-1/genetics , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Middle Aged , Nucleus Pulposus/cytology , Nucleus Pulposus/metabolism , Transcription Factor RelA/antagonists & inhibitors , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , Young Adult , beta Catenin/metabolism
6.
Biochim Biophys Acta Biomembr ; 1861(7): 1302-1316, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31077676

ABSTRACT

Aggregated LDL is the first ligand reported to interact with the cluster II CR9 domain of low-density lipoprotein receptor-related protein 1 (LRP1). In particular, the C-terminal half of domain CR9, comprising the region Gly1127-Cys1140 exclusively recognizes aggregated LDL and it is crucial for aggregated LDL binding. Our aim was to study the effect of the sequence Gly1127-Cys1140 (named peptide LP3 and its retro-enantio version, named peptide DP3) on the structural characteristics of sphingomyelinase- (SMase) and phospholipase 2 (PLA2)-modified LDL particles. Turbidimetry, gel filtration chromatography (GFC) and transmission electronic microscopy (TEM) analysis showed that LP3 and DP3 peptides strongly inhibited SMase- and PLA2-induced LDL aggregation. Nondenaturing polyacrylamide gradient gel electrophoresis (GGE), agarose gel electrophoresis and high-performance thin-layer chromatography (HPTLC) indicated that LP3 and DP3 prevented SMase-induced alterations in LDL particle size, electric charge and phospholipid content, respectively, but not those induced by PLA2. Western blot analysis showed that LP3 and DP3 counteracted changes in ApoB-100 conformation induced by the two enzymes. LDL proteomics (LDL trypsin digestion followed by mass spectroscopy) and computational modeling methods evidenced that peptides preserve ApoB-100 conformation due to their electrostatic interactions with a basic region of ApoB-100. These results demonstrate that LRP1-derived peptides are protective against LDL aggregation, even in conditions of extreme lipolysis, through their capacity to bind to ApoB-100 regions critical for ApoB-100 conformational preservation. These results suggests that these LRP1(CR9) derived peptides could be promising tools to prevent LDL aggregation induced by the main proteolytic enzymes acting in the arterial intima.


Subject(s)
Lipoproteins, LDL/chemistry , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Peptides/metabolism , Arthropod Proteins/blood , Humans , Low Density Lipoprotein Receptor-Related Protein-1/chemistry , Oligopeptides/blood , Phospholipases A2/metabolism , Phospholipids/chemistry , Protein Binding , Sphingomyelin Phosphodiesterase/chemistry , Static Electricity
7.
J Neurovirol ; 25(5): 648-660, 2019 10.
Article in English | MEDLINE | ID: mdl-31016584

ABSTRACT

In patients infected with the human immunodeficiency virus (HIV), the HIV-Tat protein may be continually produced despite adequate antiretroviral therapy. As the HIV-infected population is aging, it is becoming increasingly important to understand how HIV-Tat may interact with proteins such as amyloid ß and Tau which accumulate in the aging brain and eventually result in Alzheimer's disease. In this review, we examine the in vivo data from HIV-infected patients and animal models and the in vitro experiments that show how protein complexes between HIV-Tat and amyloid ß occur through novel protein-protein interactions and how HIV-Tat may influence the pathways for amyloid ß production, degradation, phagocytosis, and transport. HIV-Tat may also induce Tau phosphorylation through a cascade of cellular processes that lead to the formation of neurofibrillary tangles, another hallmark of Alzheimer's disease. We also identify gaps in knowledge and future directions for research. Available evidence suggests that HIV-Tat may accelerate Alzheimer-like pathology in patients with HIV infection which cannot be impacted by current antiretroviral therapy.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , HIV Infections/metabolism , Peptide Fragments/metabolism , tat Gene Products, Human Immunodeficiency Virus/metabolism , tau Proteins/metabolism , Alzheimer Disease/etiology , Alzheimer Disease/pathology , Amyloid beta-Peptides/chemistry , Animals , Anti-HIV Agents/therapeutic use , Brain Chemistry , Extracellular Space , HIV Infections/drug therapy , Humans , Low Density Lipoprotein Receptor-Related Protein-1/chemistry , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Lysosomes/metabolism , Macrophages/metabolism , Macrophages/virology , Mice , Models, Molecular , Neprilysin/chemistry , Neprilysin/metabolism , Neurofibrillary Tangles/metabolism , Neuroglia/metabolism , Neuroglia/virology , Peptide Fragments/chemistry , Phagocytosis , Phosphorylation , Protein Binding , Protein Conformation , Protein Interaction Mapping , Protein Processing, Post-Translational , Protein Transport , Structure-Activity Relationship , tat Gene Products, Human Immunodeficiency Virus/chemistry , tat Gene Products, Human Immunodeficiency Virus/genetics , tat Gene Products, Human Immunodeficiency Virus/toxicity , tau Proteins/chemistry
8.
Mol Neurobiol ; 56(10): 7234-7245, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31004319

ABSTRACT

The low-density lipoprotein receptor-related protein-1 (LRP1) has a dual role in the metabolism of the amyloid precursor protein (APP). In cellular models, LRP1 enhances amyloid-ß (Aß) generation via APP internalization and thus its amyloidogenic processing. However, conditional knock-out studies in mice define LRP1 as an important mediator for the clearance of extracellular Aß from brain via cellular degradation or transcytosis across the blood-brain barrier (BBB). In order to analyze the net effect of LRP1 on production and clearance of Aß in vivo, we crossed mice with impaired LRP1 function with a mouse model of Alzheimer's disease (AD). Analysis of Aß metabolism showed that, despite reduced Aß clearance due to LRP1 inactivation in vivo, less Aß was found in cerebrospinal fluid (CSF) and brain interstitial fluid (ISF). Further analysis of APP metabolism revealed that impairment of LRP1 in vivo shifted APP processing from the Aß-generating amyloidogenic cleavage by beta-secretase to the non-amyloidogenic processing by alpha-secretase as shown by a decrease in extracellular Aß and an increase of soluble APP-α (sAPP-α). This shift in APP processing resulted in overall lower Aß levels and a reduction in plaque burden. Here, we present for the first time clear in vivo evidence that global impairment of LRP1's endocytosis function favors non-amyloidogenic processing of APP due to its reduced internalization and subsequently, reduced amyloidogenic processing. By inactivation of LRP1, the inhibitory effect on Aß generation overrules the simultaneous impaired Aß clearance, resulting in less extracellular Aß and reduced plaque deposition in a mouse model of AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Amino Acid Motifs , Animals , Brain/metabolism , Disease Models, Animal , Endothelial Cells/metabolism , Low Density Lipoprotein Receptor-Related Protein-1/chemistry , Mice , Mutation/genetics , Plaque, Amyloid/metabolism
9.
Drug Deliv Transl Res ; 9(1): 394-403, 2019 02.
Article in English | MEDLINE | ID: mdl-30136122

ABSTRACT

Alzheimer's disease (AD) is currently incurable and places a large burden on the caregivers of AD patients. In the AD brain, iron is abundant, catalyzing free radicals and impairing neurons. The blood-brain barrier hampers antidementia drug delivery via circulation to the brain, which limits the therapeutic effects of drugs. Here, according to the method described by Gobinda, we synthesized a 16 lysine (K) residue-linked low-density lipoprotein receptor-related protein (LRP)-binding amino acid segment of apolipoprotein E (K16APoE). By mixing this protein with our designed therapeutic peptide HAYED, we successfully transported HAYED into an AD model mouse brain, and the peptide scavenged excess iron and radicals and decreased the necrosis of neurons, thus easing AD.


Subject(s)
Alzheimer Disease/drug therapy , Apolipoproteins E/chemistry , Low Density Lipoprotein Receptor-Related Protein-1/chemistry , Peptides/administration & dosage , Animals , Apolipoproteins E/metabolism , Biological Transport , Blood-Brain Barrier/drug effects , Disease Models, Animal , Humans , Iron/chemistry , Mice , Peptides/chemistry
10.
Autophagy ; 15(2): 242-258, 2019 02.
Article in English | MEDLINE | ID: mdl-30176157

ABSTRACT

Helicobacter pylori-derived CagA, a type IV secretion system effector, plays a role as an oncogenic driver in gastric epithelial cells. However, upon delivery into gastric epithelial cells, CagA is usually degraded by macroautophagy/autophagy. Hence, the induction of autophagy in H. pylori-infected epithelial cells is an important host-protective ability against gastric carcinogenesis. However, the mechanisms by which autophagosome-lysosome fusion is regulated, are unknown. Here, we report that enhancement of LAMP1 (lysosomal associated membrane protein 1) expression is necessary for autolysosome formation. LAMP1 expression is induced by nuclear translocated LRP1 (LDL receptor related protein 1) intracellular domain (LRP1-ICD) binding to the proximal LAMP1 promoter region. Nuclear translocation of LRP1-ICD is enhanced by H. pylori infection. In contrast, CAPZA1 (capping actin protein of muscle Z-line alpha subunit 1) inhibits LAMP1 expression via binding to LRP1-ICD in the nuclei. The binding of CAPZA1 to LRP1-ICD prevents LRP1-ICD binding to the LAMP1 proximal promoter. Thus, in CAPZA1-overexpressing gastric epithelial cells infected with H. pylori, autolysosome formation is inhibited and CagA escapes autophagic degradation. These findings identify CAPZA1 as a novel negative regulator of autolysosome formation and suggest that deregulation of CAPZA1 expression leads to increased risk of gastric carcinogenesis. Abbreviations: CagA: cytotoxin-associated gene A; CAPZA1: capping actin protein of muscle Z-line alpha subunit 1; ChIP: chromatin immunoprecipitation; GTF2I: general transcription factor IIi; HDAC: histone deacetylase; LAMP1: lysosomal associated membrane protein 1; LRP1: LDL receptor related protein 1; LRP1-ICD: CagA intracellular domain; qPCR: quantitative polymerase chain reaction; VacA: vacuolating cytotoxin.


Subject(s)
Antigens, Bacterial/metabolism , Autophagy , Bacterial Proteins/metabolism , CapZ Actin Capping Protein/metabolism , Carcinogenesis/pathology , Helicobacter pylori/metabolism , Proteolysis , Stomach Neoplasms/metabolism , Autophagy/drug effects , Base Sequence , CapZ Actin Capping Protein/genetics , Carcinogenesis/drug effects , Carcinogenesis/metabolism , Cell Line, Tumor , Gastric Mucosa/drug effects , Gastric Mucosa/microbiology , Gastric Mucosa/pathology , Helicobacter Infections/metabolism , Helicobacter Infections/pathology , Helicobacter pylori/drug effects , Histone Deacetylase Inhibitors/pharmacology , Humans , Low Density Lipoprotein Receptor-Related Protein-1/chemistry , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Lysosomal Membrane Proteins/metabolism , Lysosomes/drug effects , Lysosomes/metabolism , Models, Biological , Protein Binding/drug effects , Protein Domains , Proteolysis/drug effects , Risk Factors , Stomach Neoplasms/genetics , Stomach Neoplasms/microbiology , Stomach Neoplasms/pathology
11.
PLoS One ; 12(6): e0180354, 2017.
Article in English | MEDLINE | ID: mdl-28662213

ABSTRACT

BACKGROUND: Low density lipoprotein (LDL) receptor-related protein 1 (LRP1) is a ubiquitously expressed multi-ligand endocytosis receptor implicated in a wide range of signalling, among others in tumour biology. Tumour-associated genomic mutations of the LRP1 gene are described, but nothing is known about cancer-associated expression of LRP1 splice variants Therefore, the focus of this study was on an annotated truncated LRP1 splice variant (BC072015.1; NCBI GenBank), referred to as smLRP1, which was initially identified in prostate and lung carcinoma. METHODS: Using PCR and quantitative PCR, the expression of LRP1 and smLRP1 in different human tissues and tumour cell lines was screened and compared on tumour biopsies of head and neck squamous cell carcinoma (HNSCC). Using a recently developed anti-smLRP1 antibody, the expression of the putative LRP1 protein isoform in tumour cell lines in Western blot and immunofluorescence staining was further investigated. RESULTS: The alternative transcript smLRP1 is ubiquitously expressed in 12 human cell lines of different origin and 22 tissues which is similar to LRP1. A shift in expression of smLRP1 relative to LRP1 towards smLRP1 was observed in most tumour cell lines compared to healthy tissue. The expression of LRP1 as well as smLRP1 is decreased in HNSCC cell lines in comparison to healthy mucosa. In vitro results were checked using primary HNSCC. Furthermore, the expression of the protein isoform smLRP1 (32 kDa) was confirmed in human tumour cell lines. CONCLUSIONS: Similar to LRP1, the truncated splice variant smLRP1 is ubiquitously expressed in healthy human tissues, but altered in tumours pointing to a potential role of smLRP1 in cancer. Comparative results suggest a shift in expression in favour of smLRP1 in tumour cells that warrant further evaluation. The protein isoform is suggested to be secreted.


Subject(s)
Alternative Splicing , Low Density Lipoprotein Receptor-Related Protein-1/genetics , Amino Acid Sequence , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Head and Neck Neoplasms/pathology , Humans , Low Density Lipoprotein Receptor-Related Protein-1/chemistry , Sequence Homology, Amino Acid
12.
Biochim Biophys Acta Gen Subj ; 1861(8): 2088-2098, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28552561

ABSTRACT

BACKGROUND: Factor XII (FXII) is a serine protease that is involved in activation of the intrinsic blood coagulation, the kallikrein-kinin system and the complement cascade. Although the binding of FXII to the cell surface has been demonstrated, the consequence of this event for proteolytic processing of membrane-anchored proteins has never been described. METHODS: The effect of FXII on the proteolytic processing of the low-density lipoprotein receptor-related protein 1 (LRP1) ectodomain was tested in human primary lung fibroblasts (hLF), alveolar macrophages (hAM) and in human precision cut lung slices (hPCLS). The identity of generated LRP1 fragments was confirmed by MALDI-TOF-MS. Activity of FXII and gelatinases was measured by S-2302 hydrolysis and zymography, respectively. RESULTS: Here, we demonstrate a new function of FXII, namely its ability to process LRP1 extracellular domain. Incubation of hLF, hAM, or hPCLS with FXII resulted in the accumulation of LRP1 ectodomain fragments in conditioned media. This effect was independent of metalloproteases and required FXII proteolytic activity. Binding of FXII to hLF surface induced its conversion to FXIIa and protected FXIIa against inactivation by a broad spectrum of serine protease inhibitors. Preincubation of hLF with collagenase I impaired FXII activation and, in consequence, LRP1 cleavage. FXII-triggered LRP1 processing was associated with the accumulation of gelatinases (MMP-2 and MMP-9) in conditioned media. CONCLUSIONS: FXII controls LRP1 levels and function at the plasma membrane by modulating processing of its ectodomain. GENERAL SIGNIFICANCE: FXII-dependent proteolytic processing of LRP1 may exacerbate extracellular proteolysis and thus promote pathological tissue remodeling.


Subject(s)
Factor XII/pharmacology , Low Density Lipoprotein Receptor-Related Protein-1/chemistry , Gelatinases/metabolism , Humans , Protein Domains , Proteolysis
13.
J Lipid Res ; 58(7): 1267-1281, 2017 07.
Article in English | MEDLINE | ID: mdl-28381441

ABSTRACT

Among the LDL receptor (LDLR) family members, the roles of LDLR-related protein (LRP)1 in the pathogenesis of Alzheimer's disease (AD), especially late-onset AD, have been the most studied by genetic, neuropathological, and biomarker analyses (clinical studies) or cellular and animal model systems (preclinical studies) over the last 25 years. Although there are some conflicting reports, accumulating evidence from preclinical studies indicates that LRP1 not only regulates the metabolism of amyloid-ß peptides (Aßs) in the brain and periphery, but also maintains brain homeostasis, impairment of which likely contributes to AD development in Aß-independent manners. Several preclinical studies have also demonstrated an involvement of LRP1 in regulating the pathogenic role of apoE, whose gene is the strongest genetic risk factor for AD. Nonetheless, evidence from clinical studies is not sufficient to conclude how LRP1 contributes to AD development. Thus, despite very promising results from preclinical studies, the role of LRP1 in AD pathogenesis remains to be further clarified. In this review, we discuss the potential mechanisms underlying how LRP1 affects AD pathogenesis through Aß-dependent and -independent pathways by reviewing both clinical and preclinical studies. We also discuss potential therapeutic strategies for AD by targeting LRP1.


Subject(s)
Alzheimer Disease/etiology , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Apolipoproteins E/metabolism , Biomarkers/chemistry , Biomarkers/metabolism , Humans , Low Density Lipoprotein Receptor-Related Protein-1/chemistry , Molecular Targeted Therapy
14.
J Lipid Res ; 58(6): 1036-1043, 2017 06.
Article in English | MEDLINE | ID: mdl-28292942

ABSTRACT

The LDL receptor (LDLR) family has long been studied for its role in cholesterol transport and metabolism; however, the identification of ApoE4, an LDLR ligand, as a genetic risk factor for late-onset Alzheimer's disease has focused attention on the role this receptor family plays in the CNS. Surprisingly, it was discovered that two LDLR family members, ApoE receptor 2 (Apoer2) and VLDL receptor (Vldlr), play key roles in brain development and adult synaptic plasticity, primarily by mediating Reelin signaling. This review focuses on Apoer2 and Vldlr signaling in the CNS and its role in human disease.


Subject(s)
Central Nervous System Diseases/metabolism , Central Nervous System/metabolism , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Receptors, LDL/metabolism , Alternative Splicing , Animals , Central Nervous System/pathology , Central Nervous System/physiology , Central Nervous System/physiopathology , Central Nervous System Diseases/pathology , Central Nervous System Diseases/physiopathology , Central Nervous System Diseases/therapy , Humans , Low Density Lipoprotein Receptor-Related Protein-1/chemistry , Low Density Lipoprotein Receptor-Related Protein-1/genetics , Receptors, LDL/chemistry , Reelin Protein , Signal Transduction
15.
J Biol Chem ; 292(3): 912-924, 2017 01 20.
Article in English | MEDLINE | ID: mdl-27956551

ABSTRACT

LDL receptor-related protein 1 (LRP1) is a highly modular protein and the largest known mammalian endocytic receptor. LRP1 binds and internalizes many plasma components, playing multiple crucial roles as a scavenger and signaling molecule. One major challenge to studying LRP1 has been that it is difficult to express such a large, highly glycosylated, and cysteine-rich protein, limiting structural studies to LRP1 fragments. Here, we report the first recombinant expression of the complete 61 domains of the full-length LRP1 ectodomain. This advance was achieved with a multistep cloning approach and by using DNA dilutions to improve protein yields. We investigated the binding properties of LRP1 using receptor-associated protein (RAP) as a model ligand due to its tight binding interaction. The LRP1 conformation was studied in its bound and unbound state using mass spectrometry, small-angle X-ray scattering, and negative-stain electron microscopy at neutral and acidic pH. Our findings revealed a pH-dependent release of the ligand associated with a conformational change of the receptor. In summary, this investigation of the complete LRP1 ectodomain significantly advances our understanding of this important receptor and provides the basis for further elucidating the mechanism of action of LRP1 in a whole and integrated system.


Subject(s)
Low Density Lipoprotein Receptor-Related Protein-1/chemistry , Glycosylation , HEK293 Cells , Humans , Hydrogen-Ion Concentration , Low Density Lipoprotein Receptor-Related Protein-1/genetics , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Protein Domains , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Structure-Activity Relationship , X-Ray Diffraction
16.
J Biol Chem ; 291(50): 26035-26044, 2016 Dec 09.
Article in English | MEDLINE | ID: mdl-27794518

ABSTRACT

Hemophilia A is a bleeding disorder caused by a deficiency in coagulation factor VIII (fVIII) that affects 1 in 5,000 males. Current prophylactic replacement therapy, although effective, is difficult to maintain due to the cost and frequency of injections. Hepatic clearance of fVIII is mediated by the LDL receptor-related protein 1 (LRP1), a member of the LDL receptor family. Although it is well established that fVIII binds LRP1, the molecular details of this interaction are unclear as most of the studies have been performed using fragments of fVIII and LRP1. In the current investigation, we examine the binding of intact fVIII to full-length LRP1 to gain insight into the molecular interaction. Chemical modification studies confirm the requirement for lysine residues in the interaction of fVIII with LRP1. Examination of the ionic strength dependence of the interaction of fVIII with LRP1 resulted in a Debye-Hückel plot with a slope of 1.8 ± 0.5, suggesting the involvement of two critical charged residues in the interaction of fVIII with LRP1. Kinetic studies utilizing surface plasmon resonance techniques reveal that the high affinity of fVIII for LRP1 results from avidity effects mediated by the interactions of two sites in fVIII with complementary sites on LRP1 to form a bivalent fVIII·LRP1 complex. Furthermore, although fVIII bound avidly to soluble forms of clusters II and IV from LRP1, only soluble cluster IV competed with the binding of fVIII to full-length LRP1, revealing that cluster IV represents the major fVIII binding site in LRP1.


Subject(s)
Factor VIII/metabolism , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Multiprotein Complexes/metabolism , Animals , Binding Sites , Cell Line , Cricetinae , Factor VIII/chemistry , Factor VIII/genetics , Humans , Kinetics , Low Density Lipoprotein Receptor-Related Protein-1/chemistry , Low Density Lipoprotein Receptor-Related Protein-1/genetics , Male , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Surface Plasmon Resonance
17.
Biomacromolecules ; 17(10): 3252-3261, 2016 10 10.
Article in English | MEDLINE | ID: mdl-27610455

ABSTRACT

Sulfated glycosaminoglycans (sGAGs) modulate cellular processes via their interaction with extracellular matrix (ECM) proteins. We revealed a direct binding of tissue inhibitor of metalloproteinase-3 (TIMP-3) to the endocytic receptor low-density lipoprotein receptor-related protein (LRP-1) clusters II and IV using surface plasmon resonance. Sulfated hyaluronan (sHA) and chondroitin sulfate (sCS) derivatives interfered with TIMP-3/LRP-1 complex formation in a sulfation-dependent manner stronger than heparin. Electrostatic potential calculations suggested a competition between negatively charged GAGs and highly negatively charged complement-like domains of LRP-1 for the binding to a positively charged area of TIMP-3 as an underlying mechanism. In vitro studies revealed increased amounts of pericellular TIMP-3 in the presence of sHA as a consequence of the blocked protein uptake. GAG derivatives as part of biomaterials might post-translationally modulate TIMP-3 levels stronger than native GAGs, thus exhibiting catabolic effects on the ECM, which could prevent extensive pathological matrix degradation and promote wound healing.


Subject(s)
Glycosaminoglycans/administration & dosage , Hyaluronic Acid/administration & dosage , Low Density Lipoprotein Receptor-Related Protein-1/biosynthesis , Tissue Inhibitor of Metalloproteinase-3/biosynthesis , Chondroitin Sulfates/administration & dosage , Chondroitin Sulfates/chemistry , Endocytosis/drug effects , Gene Expression Regulation/drug effects , Glycosaminoglycans/chemistry , Humans , Hyaluronic Acid/chemistry , Low Density Lipoprotein Receptor-Related Protein-1/chemistry , Mesenchymal Stem Cells/drug effects , Protein Binding/drug effects , Surface Plasmon Resonance , Tissue Inhibitor of Metalloproteinase-3/chemistry , Wound Healing/drug effects
18.
J Biol Chem ; 291(35): 18430-9, 2016 08 26.
Article in English | MEDLINE | ID: mdl-27402839

ABSTRACT

The LDL receptor-related protein 1 (LRP1) is a large endocytic receptor that binds and mediates the endocytosis of numerous structurally diverse ligands. Currently, the basis for ligand recognition by LRP1 is not well understood. LRP1 requires a molecular chaperone, termed the receptor-associated protein (RAP), to escort the newly synthesized receptor from the endoplasmic reticulum to the Golgi. RAP is a three-domain protein that contains the following two high affinity binding sites for LRP1: one is located within domains 1 and 2, and one is located in its third domain. Studies on the interaction of the RAP third domain with LRP1 reveal critical contributions by lysine 256 and lysine 270 for this interaction. From these studies, a model for ligand recognition by this class of receptors has been proposed. Here, we employed surface plasmon resonance to investigate the binding of RAP D1D2 to LRP1. Our results reveal that the high affinity of D1D2 for LRP1 results from avidity effects mediated by the simultaneous interactions of lysine 60 in D1 and lysine 191 in D2 with sites on LRP1 to form a bivalent D1D2-LRP1 complex. When lysine 60 and 191 are both mutated to alanine, the binding of D1D2 to LRP1 is ablated. Our data also reveal that D1D2 is able to bind to a second distinct site on LRP1 to form a monovalent complex. The studies confirm the canonical model for ligand recognition by this class of receptors, which is initiated by pairs of lysine residues that dock into acidic pockets on the receptor.


Subject(s)
LDL-Receptor Related Protein-Associated Protein/chemistry , Low Density Lipoprotein Receptor-Related Protein-1/chemistry , Models, Molecular , Multiprotein Complexes/chemistry , Humans , LDL-Receptor Related Protein-Associated Protein/genetics , LDL-Receptor Related Protein-Associated Protein/metabolism , Low Density Lipoprotein Receptor-Related Protein-1/genetics , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Lysine/chemistry , Lysine/genetics , Lysine/metabolism , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Protein Binding , Protein Domains
19.
Matrix Biol ; 56: 57-73, 2016 12.
Article in English | MEDLINE | ID: mdl-27084377

ABSTRACT

Matrix metalloproteinase 13 (MMP-13) degrades collagenous extracellular matrix and its aberrant activity associates with diseases such as arthritis, cancer, atherosclerosis and fibrosis. The wide range of MMP-13 proteolytic capacity suggests that it is a powerful, potentially destructive proteinase and thus it has been believed that MMP-13 is not produced in most adult human tissues in the steady state. Present study has revealed that human chondrocytes isolated from healthy adults constitutively express and secrete MMP-13, but that it is rapidly endocytosed and degraded by chondrocytes. Both pro- and activated MMP-13 bind to clusters II and III of low-density lipoprotein (LDL) receptor-related protein 1 (LRP1). Domain deletion studies indicated that the hemopexin domain is responsible for this interaction. Binding competition between MMP-13 and ADAMTS-4, -5 or TIMP-3, which also bind to cluster II, further shown that the MMP-13 binding site within cluster II is different from those of ADAMTS-4, -5 or TIMP-3. MMP-13 is therefore co-endocytosed with ADAMTS-5 and TIMP-3 by human chondrocytes. These findings indicate that MMP-13 may play a role on physiological turnover of cartilage extracellular matrix and that LRP1 is a key modulator of extracellular levels of MMP-13 and its internalization is independent of the levels of ADAMTS-4, -5 and TIMP-3.


Subject(s)
ADAMTS5 Protein/metabolism , Chondrocytes/enzymology , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Matrix Metalloproteinase 13/metabolism , Tissue Inhibitor of Metalloproteinase-3/metabolism , ADAMTS5 Protein/chemistry , Binding, Competitive , Endocytosis , HEK293 Cells , Humans , Low Density Lipoprotein Receptor-Related Protein-1/chemistry , Matrix Metalloproteinase 13/chemistry , Protein Binding , Protein Interaction Domains and Motifs , Protein Transport , Tissue Inhibitor of Metalloproteinase-3/chemistry
20.
J Biol Chem ; 290(24): 14852-65, 2015 Jun 12.
Article in English | MEDLINE | ID: mdl-25918169

ABSTRACT

Low density lipoprotein receptor-related protein (LRP1) mediates the internalization of aggregated LDL (AgLDL), which in turn increases the expression of LRP1 in human vascular smooth muscle cells (hVSMCs). This positive feedback mechanism is thus highly efficient to promote the formation of hVSMC foam cells, a crucial vascular component determining the susceptibility of atherosclerotic plaque to rupture. Here we have determined the LRP1 domains involved in AgLDL recognition with the aim of specifically blocking AgLDL internalization in hVSMCs. The capacity of fluorescently labeled AgLDL to bind to functional LRP1 clusters was tested in a receptor-ligand fluorometric assay made by immobilizing soluble LRP1 "minireceptors" (sLRP1-II, sLRP1-III, and sLRP1-IV) recombinantly expressed in CHO cells. This assay showed that AgLDL binds to cluster II. We predicted three well exposed and potentially immunogenic peptides in the CR7-CR9 domains of this cluster (termed P1 (Cys(1051)-Glu(1066)), P2 (Asp(1090)-Cys(1104)), and P3 (Gly(1127)-Cys(1140))). AgLDL, but not native LDL, bound specifically and tightly to P3-coated wells. Rabbit polyclonal antibodies raised against P3 prevented AgLDL uptake by hVSMCs and were almost twice as effective as anti-P1 and anti-P2 Abs in reducing intracellular cholesteryl ester accumulation. Moreover, anti-P3 Abs efficiently prevented AgLDL-induced LRP1 up-regulation and counteracted the down-regulatory effect of AgLDL on hVSMC migration. In conclusion, domain CR9 appears to be critical for LRP1-mediated AgLDL binding and internalization in hVSMCs. Our results open new avenues for an innovative anti-VSMC foam cell-based strategy for the treatment of vascular lipid deposition in atherosclerosis.


Subject(s)
Foam Cells/cytology , Lipoproteins, LDL/physiology , Low Density Lipoprotein Receptor-Related Protein-1/physiology , Muscle, Smooth, Vascular/cytology , Amino Acid Sequence , Animals , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Humans , Ligands , Low Density Lipoprotein Receptor-Related Protein-1/chemistry , Molecular Sequence Data , Real-Time Polymerase Chain Reaction , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...